+ All Categories
Home > Documents > Mir Farshid Alemdehy & Stefan J. Erkeland › pub › 75009 › REPUB_75009_OA.pdfMir Farshid...

Mir Farshid Alemdehy & Stefan J. Erkeland › pub › 75009 › REPUB_75009_OA.pdfMir Farshid...

Date post: 28-Jan-2021
Category:
Upload: others
View: 5 times
Download: 0 times
Share this document with a friend
10
Full Terms & Conditions of access and use can be found at http://www.tandfonline.com/action/journalInformation?journalCode=kccy20 Download by: [Erasmus University] Date: 17 June 2016, At: 00:53 Cell Cycle ISSN: 1538-4101 (Print) 1551-4005 (Online) Journal homepage: http://www.tandfonline.com/loi/kccy20 Stop the dicing in hematopoiesis: What have we learned? Mir Farshid Alemdehy & Stefan J. Erkeland To cite this article: Mir Farshid Alemdehy & Stefan J. Erkeland (2012) Stop the dicing in hematopoiesis: What have we learned?, Cell Cycle, 11:15, 2799-2807, DOI: 10.4161/cc.21077 To link to this article: http://dx.doi.org/10.4161/cc.21077 Copyright © 2012 Landes Bioscience Published online: 01 Aug 2012. Submit your article to this journal Article views: 82 View related articles Citing articles: 5 View citing articles
Transcript
  • Full Terms & Conditions of access and use can be found athttp://www.tandfonline.com/action/journalInformation?journalCode=kccy20

    Download by: [Erasmus University] Date: 17 June 2016, At: 00:53

    Cell Cycle

    ISSN: 1538-4101 (Print) 1551-4005 (Online) Journal homepage: http://www.tandfonline.com/loi/kccy20

    Stop the dicing in hematopoiesis: What have welearned?

    Mir Farshid Alemdehy & Stefan J. Erkeland

    To cite this article: Mir Farshid Alemdehy & Stefan J. Erkeland (2012) Stop the dicing inhematopoiesis: What have we learned?, Cell Cycle, 11:15, 2799-2807, DOI: 10.4161/cc.21077

    To link to this article: http://dx.doi.org/10.4161/cc.21077

    Copyright © 2012 Landes Bioscience

    Published online: 01 Aug 2012.

    Submit your article to this journal

    Article views: 82

    View related articles

    Citing articles: 5 View citing articles

    http://www.tandfonline.com/action/journalInformation?journalCode=kccy20http://www.tandfonline.com/loi/kccy20http://www.tandfonline.com/action/showCitFormats?doi=10.4161/cc.21077http://dx.doi.org/10.4161/cc.21077http://www.tandfonline.com/action/authorSubmission?journalCode=kccy20&page=instructionshttp://www.tandfonline.com/action/authorSubmission?journalCode=kccy20&page=instructionshttp://www.tandfonline.com/doi/mlt/10.4161/cc.21077http://www.tandfonline.com/doi/mlt/10.4161/cc.21077http://www.tandfonline.com/doi/citedby/10.4161/cc.21077#tabModulehttp://www.tandfonline.com/doi/citedby/10.4161/cc.21077#tabModule

  • © 2012 Landes Bioscience.

    Do not distribute.

    www.landesbioscience.com Cell Cycle 2799

    Cell Cycle 11:15, 2799-2807; August 1, 2012; © 2012 Landes Bioscience

    extrA view extrA view

    Keywords: Dicer1, miRNA, myelopoi-esis, leukemia, hematopoietic stem cell

    Submitted: 06/01/12

    Accepted: 06/09/12

    http://dx.doi.org/10.4161/cc.21077

    *Correspondence to: Stefan J. Erkeland; Email: [email protected]

    MicroRNAs (miRNAs) belong to an abundant class of highly con-served small (22nt) non-coding RNAs. MiRNA profiling studies indicate that their expression is highly cell type-depen-dent. DICER1 is an essential RNase III endoribonuclease for miRNA process-ing. Hematopoietic cell type- and devel-opmental stage-specific Dicer1 deletion models show that miRNAs are essential regulators of cellular survival, differ-entiation and function. For instance, miRNA deficiency in hematopoietic stem cells and progenitors of different origins results in decreased cell survival, dra-matic developmental aberrations or dys-functions in mice. We recently found that homozygous Dicer1 deletion in myeloid-committed progenitors results in an aber-rant expression of stem cell genes and induces a regained self-renewal capac-ity. Moreover, Dicer1 deletion causes a block in macrophage development and myeloid dysplasia, a cellular condition that may be considered as a preleukemic state. However, Dicer1-null cells do not develop leukemia in mice, indicating that depletion of miRNAs is not enough for tumorigenesis. Surprisingly, we found that heterozygous Dicer1 deletion in myeloid-committed progenitors, but not Dicer1 knockout, collaborates with p53 deletion in leukemic progression and results in various types of leukemia. Our data indicate that Dicer1 is a haploinsuf-ficient tumorsuppressor in hematopoietic neoplasms, which is consistent with the observed downregulation of miRNA expression in human leukemia samples. Here, we review the various hematopoi-etic specific Dicer1 deletion mouse models

    Stop the dicing in hematopoiesisWhat have we learned?

    Mir Farshid Alemdehy and Stefan J. Erkeland*Department of Hematology; Erasmus University Medical Center; Rotterdam, The Netherlands

    and the phenotypes observed within the different hematopoietic lineages and cell developmental stages. Finally, we dis-cuss the role for DICER1 in mouse and human malignant hematopoiesis.

    Introduction

    DICER1 is an evolutionarily conserved member of the RNase III family of endori-bonucleases. The gene encoding DICER1 is located on human chromosome 14q32 and mouse chromosome 12E. DICER1 is a complex protein and contains three N-terminal Helicase domains (HEL1, HEL2i, HEL2), a DUF283 domain, which is presumably involved in bind-ing of double-stranded RNA (dsRNA), a Platform domain, the pre-miRNA bind-ing domain PAZ, RNase IIIa, RNase IIIb and a C-terminal dsRNA binding domain (dsRBD).1-3 The RNase III domains of DICER1 cleave double-stranded RNA (dsRNA) substrates and specific precursor hairpin sequences, including so-called pre-miRNAs, into small 5'-phosphorylated RNAs of typically 21–23 nucleotides called miRNA.4 Deep sequencing of 5'-phos-phorylated short RNAs in ES cells showed that the miRNA is the only class of short RNAs to be fully DICER1-dependent.5 However, the premature miR-451 is the single well-conserved miRNA-containing sequence known to bypass DICER1 pro-cessing and is matured by an Argonaute-2 (Ago-2)-dependent mechanism.6-9 The DICER1-generated short RNAs bind to Argonaute proteins in the so-called RNA-induced silencing complex (RISC). This complex induces degradation or inhibits translation of homologs target mRNAs.

    Dow

    nloa

    ded

    by [

    Era

    smus

    Uni

    vers

    ity]

    at 0

    0:53

    17

    June

    201

    6

  • © 2012 Landes Bioscience.

    Do not distribute.

    2800 Cell Cycle volume 11 issue 15

    Tregs, using a Foxp3-Cre knock-in mouse (Fig. 1C).21,22 Under steady-state condi-tions, Foxp3-controlled deletion of Dicer1 has minimal effects on Treg cell develop-ment, cellular proliferation and survival in the peripheral compartments.22 However, a diminished fitness of Dicer1-deficient Treg cells in the periphery was observed in a competitive experiment in mice.21 Under inflammatory conditions, the immune-repressive capacity of the mutant Treg cells is markedly reduced and results in rapid fatal autoimmunity and complete failure of immune suppression activity.21,22 Moreover, Dicer1 deletion in Treg cells leads to the progression of fatal lympho-proliferative autoimmune syndrome with an early onset, which is indistinguish-ably comparable to T-cell-specific Foxp3 deficiency.21 The expression of putative suppressor effector molecules, includ-ing CTLA4, IL-10, EBV-induced gene 3 (Ebi-3) and granzyme B, was decreased by still-unidentified miRNA-controlled mechanisms.21 Tregs express a specific set of miRNAs, including miR-223, miR-155 and miR-146, which is distinct from naïve CD4+ T-cells.20 Therefore, the expression of these miRNAs may be under direct or indirect control of the transcription fac-tor Foxp3.20 Identification of the targets that are controlled by these miRNAs in Tregs may provide new insights about the molecular pathways involved in the activ-ity of these cells.

    The role of miRNAs in invariant Natural Killer T (iNKT) cells was studied in a mouse strain by Tie2-Cre-mediated disruption of Dicer1.23 The Tie2 kinase is specifically expressed in hematopoi-etic progenitors and endothelial cells.24 Similar to the immune phenotypes in CD4-Cre;Dicer1fl/fl and Lck-Cre;Dicer1fl/ fl, these mice show reduced numbers of iNKT cells in the thymus, spleen and liver. Moreover, Dicer1 deletion results in devel-opmental abnormalities of iNKT cells.23,25 In addition, Dicer1-deficient peripheral iNKT cell numbers are decreased and displayed profound defects in α-GalCer, phorbol myristate acetate (PMA) and ionomycin-induced cellular activation and production of cytokines such as IL-4 and IFN-γ.23 Together, these data indicate that Dicer1 controls survival at the early T-cell developmental stage. At the later

    to be essential for the generation and sur-vival of αβ T-cells. However, in the sur-viving T-cells, Dicer1 is dispensable for CD4+ and CD8+ single positive lineage commitment.18 These results strongly sug-gest that Dicer1 deletion does not affect normal T-cell lineage-specific gene expres-sion programs. In these cells, the transcrip-tional repression of centromeric satellite repeats and features of facultative hetero-chromatin are maintained in the absence of Dicer1,18 suggesting that survival of immature T-cells is regulated directly by a miRNA-controlled mechanism.

    The CD4-Cre transgenic mouse model enables investigation of the consequences of Dicer1 deletion at a later stage of T cell development (Fig. 1B). These mice show four major phenotypes: (1) Dicer1 is required for basic cellular processes, such as proliferation and survival, as also proposed by Cobb et al., and therefore Dicer1 deficiency results in decreased number of T-cells.18,19 (2) Dicer1 deletion appears to favor T-cell lineage production from CD4+CD8+ double-positive stage toward CD4+ single-positive peripheral T-cells over CD8+ single-positive cells. However, this phenotype was less obvious from thymic T-cell lineage analysis. This discrepancy may be explained by the fact that CD4-Cre-driven deletion of Dicer1 does not result in complete depletion of all miRNAs, presumably due to high miRNA stability and limited cell divisions of a small fraction of CD4+ T-cells, which may be different for Dicer1-null CD8+ T-cells. (3) Dicer1-null CD4+ T-cells pro-duce increased levels of IFN-γ, a pro-Th1 cytokine, indicating that Dicer1 controls Th1-lineage commitment.19 (4) CD4-Cre; Dicer1fl/fl mice show a more than 2-fold decreased proportion of Foxp3+ regula-tory T cells (Treg).20 Interestingly, these mice developed a splenomegaly, and their lymph nodes were severely enlarged at the age of 3 to 4 mo. Moreover, organs such as colon, lung and liver were affected by immune pathology caused by an overac-tive immune system, which is less severe as compared with Foxp3-knockout mice lacking functional Tregs.20 However, this phenotype suggests that Dicer1-deficient Tregs are functionally aberrant as well.

    Two studies revealed the role of Dicer1 more specifically in the function of mature

    Moreover RISC triggers gene silencing via chromatin modifications at target promot-ers under specific conditions such as cel-lular senescence.10,11

    Genetic studies in plants, zebrafish and mice show that Dicer1 is essential for nor-mal development.12-14 For instance, genetic deletion of Dicer1 in mice results in early embryonic mortality due to depletion of the Oct-4-positive pluripotent embry-onic stem cell pool at embryonic day (E) 6-E7.14 Dicer1-null ES cells are incapable of processing miRNA hairpins or dsR-NAs.5,15,16 However, Dicer1 is dispensable for the siRNA-mediated gene silencing response.16 Although a role for Dicer1 in centromeric silencing has been suggested, deep sequencing of small RNAs in Dicer1-null and Dicer1 wild type ES cells indi-cates that the production of miRNAs is the sole catalytic function of DICER1 in these cells.5 To bypass embryonic lethal-ity and to enable investigation of Dicer1 functions in adult tissues in mice, a floxed Dicer1 allele (Dicer1fl) has been generated that allows conditional deletion of Dicer1 in a cell type- and developmental stage-specific fashion.17 To address the overall role of miRNAs in the development and function of hematopoietic cells, differ-ent hematopoietic cell stage and lineage-specific conditional Dicer1 deletion strains have been used. First, we will review the phenotypic consequences of Dicer1 dele-tion at different stages of hematopoiesis and cell types. Second, we discuss what we have learned from these models about miRNA-controlled pathways in hema-topoiesis. Finally, we show evidence for Dicer1 haploinsufficient tumorsuppressor activity in mouse leukemia and discuss the role for DICER1 in human AML.

    The Role of Dicer1 in T-Lymphocyte Development

    In one of the first studies addressing the role of Dicer1 in hematopoiesis in vivo, floxed Dicer1 alleles were deleted by CRE in lymphocyte-specific protein tyrosine kinase (Lck)-positive cells. In this model, Cre is active at the double-negative (DN) CD4-CD8- T cell developmental stage and results in Dicer1-null CD44-CD25- (DN4), CD4+CD8+ and CD4+CD8-, CD4-CD8- cells (Fig. 1A).18 Dicer1 seems

    Dow

    nloa

    ded

    by [

    Era

    smus

    Uni

    vers

    ity]

    at 0

    0:53

    17

    June

    201

    6

  • © 2012 Landes Bioscience.

    Do not distribute.

    www.landesbioscience.com Cell Cycle 2801

    inhibitory genes, such as Cdkn1c (p57Kip2), Cdkn2b (p16INK4a), Cdk1a (p21Cip1) and Cdkn1b (p27Kip).29 Furthermore, Dicer1 deletion in B-cells leads to massive induc-tion of apoptosis due to derepression of the proapoptotic protein BIM1 as described for early stages of B-cell development.29 Together, these data show that Dicer1 con-trols survival of B-cells at different stages of B-cell development, regulates cellu-lar proliferation and is critical for proper B- and plasma cell functions.

    The Role for Dicer1 in NK Cell Function

    Bezman et al. induced ablation of con-ditional Dicer1 alleles with a tamoxi-fen-inducible Cre recombinase (human estrogen receptor (ERT2)-Cre) and studied the effects of miRNA depletion in NK cells.30 This non-specific model revealed a role for Dicer1 in the maintenance of

    produce high titers of autoreactive anti-bodies and as a result cause autoimmune disease in aged female mice.28 However, the miRNAs that control autoreactivity are still unidentified.

    To investigate the role for Dicer1 in antigen-activated, but not naive B cells, an activation-induced cytadine deami-nase (Aicda)-Cre-mediated Dicer1 dele-tion mouse model has been generated.29 This mouse model showed that Dicer1 is required for the production of antigen-specific high-affinity antibodies during a T-cell-dependent immune response.29 Also, the formation of germinal center B cells is drastically impaired in Dicer1-deficient mice.29 These mutant mice fail to generate memory B and long-lived plasma cells after immunization with a T cell-dependent antigen. This study pro-vides evidence for Dicer1-controlled cell proliferation of activated germinal center B-cells by strong repression of cell cycle

    stage, Dicer1 is critical for the balance of Th1/Th2 lineage production and controls functions such as immune-repression and specific cellular activity.

    Dicer1 Function During B Cell Development

    Ablation of Dicer1 in early B cell pro-genitors, mediated by the Mb1-Cre allele, which is expressed at the earliest stage of B-cell development, blocks B-cell development almost completely at the pro-B-cell (B220low, c-kit+ CD25-) to pre-B-cell (B220int, c-kit-, CD25+) transition (Fig. 1D).26 This block in B-cell develop-ment is caused by a strong induction of apoptosis and results in total depletion of B cells in the BM and the peripheral lym-phoid organs in mice.26 Gene expression profiling of Abelson virus (v-Abl)-trans-formed Dicer1-null pro-B-cells revealed that miR-142–3p and different members of the miR-17~92 family of miRNA, such as miR-17, miR-19, miR-20 and miR-92, are the most active at the pro-B-cell stage.26 Derepression of the proapop-totic protein BIM, a confirmed target of miR-17~92, was shown to be mainly responsible for the failure of the cells to respond to survival signals.26 In full agree-ment, Ventura A and colleagues have dem-onstrated that deletion of the miR-17~92 in mouse hematopoietic stem cells leads to a cell development arrest at the pro-B to pre-B transition that is highly reminiscent of what has been observed in the Dicer1-deficient mice.27

    The role of miRNAs in terminal B cell differentiation is addressed by the analy-sis of CD19-Cre driven Dicer1-deletion mouse model (Fig. 1E).28 In contrast to early MB1-Cre driven Dicer1 dele-tion, depletion of Dicer1 with CD19-Cre in immature B220+ IgM+ cells does not induce cell death and allowed analysis of the role for Dicer1 in mature B cells in peripheral tissues.28 In the absence of Dicer1, transitional and marginal zone B cells are overrepresented, and the genera-tion of follicular B cells is impaired.28 The miR-185 is abundantly expressed in fol-licular B-cells and controls the expression of B cell antigen receptor (BCR) signaling effector Bruton tyrosine kinase (BtK) in activated B cells.28 Dicer1-deficient B cells

    Figure 1. Schematic overview of the phenotypic characteristics of different Cre-mediated Dicer1-deletion models in lymphopoiesis (A) HSCs develop via different progenitors toward mature CD4+ or CD8+ single positive cells. the effects of LCK-Cre-mediated Dicer1 deletion are depicted. the apparent level of Dicer1 expression is indicated by the yellow background color (yellow, normal endogenous levels; white, no Dicer1 expression). HSC, hematopoietic stem cells; CLP, common lymphoid progenitor; DN1–3, double-negative stage 1 to 3 (CD4-CD8-); DN4, double-negative stage 4, DP: double-positive CD4+CD8+ cells. Phenotypic characteristics are indicated by the red arrows and lines. Dashed lines indicate less cells than in wild-type situation (B). See also (A). the ef-fects of CD4-Cre-mediated Dicer1 deletion are depicted. Phenotypic characteristics are indicated by the red arrow and lines (C). See also (A). the effects of FoxP3-Cre-mediated Dicer1 deletion results in normal numbers of regulatoty t-cells (tregs), but these cells are functionally aberrant. (D) HSCs develop via indicated progenitors toward mature B-cells. the effects of MB1-Cre-medi-ated Dicer1 deletion are indicated by the red lines and arrow and result in developmental block from the pro-B-cell to the pre-B-cell stage. Pro-B: earliest stage of progenitor B-cell development, pre-B-cell, precursor stage of B-cell development (e). See also (D). CD19-Cre-mediated deletion of Dicer1 results in mature B-cells which are functionally aberrant.

    Dow

    nloa

    ded

    by [

    Era

    smus

    Uni

    vers

    ity]

    at 0

    0:53

    17

    June

    201

    6

  • © 2012 Landes Bioscience.

    Do not distribute.

    2802 Cell Cycle volume 11 issue 15

    (HSPCs) was first studied by breeding Dicer1fl/fl with Mx-Cre mice.32 These mice express the Cre-recombinase in response to interferons and are highly efficient in recombination of floxed alleles in the hematopoietic system in vivo via perito-neal injection of polyI:polyC (pIpC).32 Dicer1 ablation in these mice depletes functional HSCs, induces rapid apoptosis in HSPCs and consequently causes total disruption of hematopoiesis.32 In addi-tion, miRNA-depleted HSCs are unable to reconstitute hematopoiesis in mice.32 In full agreement, Dicer1fl/fl HSCs contain-ing the VAVi-Cre transgene that is highly active in HSCs and efficient in deletion of floxed alleleles33 are incapable to recon-stitute lethally irradiated recipient mice (Erkeland SJ et al., unpublished data). Together, these data show that Dicer1 is essential for HSCs survival. Interestingly, miR-125a controls the expansion of HSCs in vivo through targeting the proapototic gene Bak1. Whether miR-125a as a single miRNA can rescue Dicer1-null HSC sur-vival and functions remains elusive, but it is more likely that multiple miRNAs are critical at this stage.

    To address the question whether miR-NAs play a role in early myeloid-lineage decisions, we deleted Dicer1 in CCAAT/enhancer-binding protein α (C/EBPA)-positive myeloid-committed progenitors in vivo (Fig. 2).34 In striking contrast to the results in HSCs and early lym-phoid progenitors, we recently found that miRNA depletion does not affect the number of myeloid-committed pro-genitor cells in mice.34 However, isolated Dicer1-deficient granulocyte-macrophage progenitors (GMPs) were defective in myeloid development and exhibited an increased self-renewal potential.34 In mice, Dicer1 deletion by C/ebpa-Cre blocked monocytic differentiation, depleted mac-rophages and myelo-dendritic cells and caused myeloid dysplasia with morpho-logical features of Pelger-Huet anomaly34 (Fig. 2). Strikingly, monocytes express low levels of proteins involved in miRNA pro-cessing and functions such as DROSHA, AGO1 and AGO2 compared with the levels found in T-cells, and are deficient for DICER1, unless the cells are forced to differentiate toward macrophages.35,36 The presence of some miRNAs in the

    as IL-15 and IL-12, tumor target cells, activating NK cell receptor ligation as well as during acute MCMV infection in vivo.31 The miR-15/16 family of miRNAs is potentially contributing to IFN-γ sup-pression and may control dampening of NK cell functions.31

    Dicer1 Deletion in Myeloid- Committed Progenitors Revealed

    an Unexpected Function in Hematopoiesis

    The consequences of Dicer1 deletion in hematopoietic stem and progenitor cells

    survival and function of NK cells.30 They found that in response to a viral infection with mouse cytomegalovirus (MCMV), the expansion of NK cells, but not the IFN-γ production, is Dicer1-dependent, suggesting that survival but not activity of NK cells is affected by Dicer1 deficiency.30 Similarly, HCD2-Cre;Dicer1fl/fl mice, which enable a lymphocyte-restricted Dicer1 deletion at the early stage of NK cells development, also showed reduced NK cell maturation and survival.30,31 However, Dicer1-null NK cells showed enhanced degranulation and IFN-γ pro-duction in response to cytokines such

    Figure 2. Schematic overview of the results of C/ebpa-Cre-mediated deletion of Dicer1 in myeloid-committed progenitors. Phenotypic characteristics are indicated in red. in short, dele-tion of Dicer1 results in derepression of stem cell genes in myeloid progenitors and an enhanced self-renewal capacity. Furthermore, MDPs and GMPs are blocked in macrophage and dendritic cell development. in addition, Dicer1 deletion results in neutrophil dysplasia with cells that are charac-teristic for Pelger-Huet anomaly. HSC/LSK, hematopoietic stem cells/Lin-;Scai+; Kit+; CMP, common myeloid progenitor; GMP, granulocyte-macrophage progenitor; MeP, megakaryocytic-erythroid progenitor; MDP, macrophage-dendritic cell progenitor; CD11B, pan marker for myeloid cells; Gr-1, marker for mature granulocytes.

    Dow

    nloa

    ded

    by [

    Era

    smus

    Uni

    vers

    ity]

    at 0

    0:53

    17

    June

    201

    6

  • © 2012 Landes Bioscience.

    Do not distribute.

    www.landesbioscience.com Cell Cycle 2803

    does not affect the viability of myeloid progenitors in mice may suggest that no negative selection due to reduced survival or proliferation by lack of miRNAs occurs in these cells. Together, these data provide evidence for a model in which reduced level of miRNAs is an oncogenic event in the development of leukemia but that activity of at least some miRNA species is essential for oncogenic transformation (Fig. 4). This is in full agreement with experimental data showing tumor sup-pressing and oncogenic activities of inves-tigated miRNAs, such as miR-17~92 and miR-125.43 Moreover, miRNA expression profiling data of human cancer and AML samples are consistent with this hypoth-esis, as a small subset of miRNAs, includ-ing e.g., miR-9, miR-125 and miR-17~92, are highly expressed, whereas most other miRNAs are downregulated.44

    Dicer1 Mutations in Human Leukemia

    To date, the mechanism behind the reduced miRNA expression in subsets of human myeloid leukemia samples still remains elusive. One possibility is that the widespread silencing of miRNAs is the result of a defect in miRNA biogenesis caused by mutations in the gene encoding DICER1. For instance, data from Cancer Genome Project at the Wellcome Trust Sanger Institute (www.sanger.ac.uk/cos-mic) show that somatic DICER1 muta-tions occur in different human tumors, including lung carcinoma, malignant melanoma and ovarian cancer.45 Recently, Hill et al. found DICER1 mutations in familial pleuro-pilmonary blastoma.46 In addition, a recent study in human non-epi-thelial ovarian cancers revealed mutations in the codons encoding metal-binding sites within the RNase IIIb catalytic cen-ters of DICER1 in 30 of 102 (29%) of the tumors.47 These authors also detected mutations in 1 out of 14 non-seminoma-tous testicular germ-cell tumors, in 2 of 5 embryonal rhabdomyosarcomas, and in 1 of 266 epithelial ovarian and endome-trial carcinomas.47 The RNase III domains of DICER1 are essential for miRNA mat-uration, and introduced mutations in the RNase IIIa and in RNase IIIb abrogate in vitro processing of the 3p and 5p miRNAs,

    not affect lymphoma latency and overall survival.41 This discrepancy may indi-cate that the tumorsuppressing activity of DICER1 is cell type-dependent.

    We asked whether Dicer1 deletion enhances myeloid leukemia development in mice. In hematopoietic cells, C/ebpa starts to be expressed in early myeloid-committed progenitors, making it a suit-able promoter to drive Dicer1 deletion for studying the role of miRNA depletion in myeloid leukemias.34,42 To circumvent pre-natal lethality, we transplanted fetal liver cells from mutant and control embryos into lethally irradiated recipient mice. While heterozygous deletion of Dicer1 in myeloid-committed progenitors does not affect myeloid development, homozygous Dicer1 deletion results in block of mac-rophage/dendritic cell development and myeloid-dysplasia, a cellular condition that may be considered as a preleukemic state34 (Fig. 2). However, mice transplanted with either heterozygous floxed Dicer1 or homo-zygous floxed Dicer1 cells survived devoid of any signs of myelo-proliferative disease or leukemia development within a year of observation, indicating that loss of Dicer1 in myeloid-committed progenitors is not sufficient to initiate short-term leukemo-genesis in mice34 (Fig. 3A). To further investigate whether depletion of miRNAs accelerates myeloid leukemia development in a tumor susceptible model, we crossed Dicer1 floxed (Dicer1fl) alleles with p53fl/fl mice and transplanted fetal liver cells from double mutants and control embryos into lethally irradiated recipient mice. C/ebpa-cre driven deletion of p53 and hemizygous deletion of Dicer1 in mice caused develop-ment of various types of leukemias in half of the reconstituted mice with a latency of approximately 6 mo (Fig. 3). Only one out of eight Dicer1f/f;p53f/f recipient mice developed a leukemia with a latency of 9 mo (Fig. 3A). However, PCR analysis on genomic DNA isolated from the Dicer1f/f tumor cells in liver and spleen showed that the Dicer1 floxed alleles were incompletely recombined (Fig. 3B). These results are in full agreement with data published by Kumar et al. and strongly suggest that only reduced levels of Dicer1, but not bialleleic loss of Dicer1, may play a functional role in leukemia development.39,40 However, the fact that total depletion of miRNAs

    monocytic and Dicer1-deficient cell line U937 suggests that some miRNAs can be generated by proteins other than DICER1, such as PIWIL4,35 but this hypothesis still needs proper validation. However, the fact that Dicer1-null monocytes are blocked in their differentiation in vivo indicates that Dicer1 is essential at this stage, and its function cannot be bypassed by other miRNA processing mechanisms.

    MiRNA profiling of wild type GMPs showed that 104 miRNAs are abundantly expressed at this stage, of which at least 20 miRNA families are potentially active by reducing their target mRNA abundance.34 Interestingly, of the derepressed miRNA targets in Dicer1-null GMPs, 27% are nor-mally exclusively expressed in HSCs or are specific for multi-potent progenitors and erythropoiesis.34 Unlike the results from HSCs and lymphoid progenitors show-ing functions of Dicer1 mainly in survival pathways, these results provide evidence for a miRNA-controlled switch of a hema-topoietic stem cell program of self-renewal and expansion toward myeloid differentia-tion (Fig. 2).34

    The Role for Dicer1 in Leukemia

    Human cancer including different types of leukemia is characterized by a global reduction in miRNA expression.37 The first experimental evidence for a role of global downregulation of miRNAs in cel-lular transformation and tumorigenesis has been presented in a K-Ras-induced mouse model for lung cancer.38 Further studies of the role for Dicer1 in human cancer devel-opment in immune-deficient mice strongly suggested that Dicer1 is a haploinsufficient tumorsuppressor.39 In this model, homo-zygous deletion of Dicer1 is tolerated by the tumor cells; however, lack of miR-NAs abrogates tumor outgrowth due to strongly reduced cell proliferation capac-ity of the DICER1-null cells.39 In agree-ment, heterozygous deletion of Dicer1, but not Dicer1-knockout, accelerated tumor formation on a retinoblastoma-sensitized background.40 In mouse B-cells, Dicer1 is required for Myc-induced B-cell lympho-magenesis and survival of B-cell lympho-mas.41 However, in this model Dicer1 is not a haploinsufficient tumor suppressor, as heterozygous deletion of Dicer1 does

    Dow

    nloa

    ded

    by [

    Era

    smus

    Uni

    vers

    ity]

    at 0

    0:53

    17

    June

    201

    6

  • © 2012 Landes Bioscience.

    Do not distribute.

    2804 Cell Cycle volume 11 issue 15

    respectively.47,48 In agreement, transient expression experiments of mutant human DICER1 constructs in murine Dicer1-null mesenchymal stem cells showed that inactivation of the RNase IIIb domain by mutation of D1709, results in complete loss of, particularly, 5p-derived mature miRNAs, including the tumor-suppres-sive Let-7 family of miRNAs.49 This muta-tion is found in subsets of nonepithelial ovarian cancers. Indeed, the identified Dicer1 hot spot mutations in cancer result in reduced RNase IIIb activity but retain RNase IIIa activity, strongly suggesting a positive selection for the mutations that reduces Let7-tumorsuppressing activity in cancer development.47

    In a first attempt to gain more func-tional insight into the mechanisms behind the reduced miRNA expression in AML, a panel of 45 AML samples, characterized by activation of the oncogene EVI-1 due to t(3;3)(q21;q26) or inv(3)(q21q26) and poor prognosis, and five AML cell lines, including U937, MOLM1, MUTZ3, KASUMI-3 and F36P, were sequenced. In this panel of high-risk AML samples, no mutation in Dicer1 coding sequences and untranslated regions were identified (unpublished data, Erkeland S.J., Valk P., Delwel H., Sanders M.A., Groschel S. and Hoogenboezem R., 2012). Despite the limited set of data, this result suggests that other mechanisms are involved in deregu-lation of miRNA expression in human AML.

    Different Mechanisms of DICER1 Activity Reduction

    in Human Leukemia

    The expression of miRNAs may be deregulated by different mechanisms in human cancer.50 For instance, the activity of DICER1 may be reduced, as DICER1 is frequently deleted in various human cancers.39 In addition, low expression of DICER1 independently predicted poor outcomes in ovarian cancer patients.51 In chronic lymphocytic leukemia (CLL), low expression of DICER1 has been cor-related with increased aggressiveness of the disease, shorter overall survival as well as reduced treatment-free survival.52 Notably, no such correlation between DICER1 transcript levels and disease

    Figure 3. Leukemia developed from C/ebpa-Cre; p53f/f; Dicer1f/wt HSCs. (A) Cumulative survival of mice transplanted with HSCs from fetal livers of C/ebpa-Cre; p53f/f; Dicer1 wt (n = 8), C/ebpa-Cre; p53f/f; Dicer1 f/wt (n = 12) and C/ebpa-Cre; p53f/f; Dicer1 f/f (n = 8) embryos. Significance: p < 0.05 (log-rank Mantel-Cox test). (B) PCr on genomic DNA extracted from tumor cells. K.O., knockout allele; fl, floxed allele; wt, wild type allele; li, liver; BM, bone marrow; spl, spleen; con, control DNA heterozygous floxed Dicer1; neg, loading control. (C) example of tumor infiltration in liver and spleen of leukemic mice transplanted with C/ebpa-Cre; p5 f/f; Dicer f/wt HSCs. (D) Micrographs show-ing morphology of tumor cells in blood, bone marrow and spleen. Bar indicates 10 μm.

    Dow

    nloa

    ded

    by [

    Era

    smus

    Uni

    vers

    ity]

    at 0

    0:53

    17

    June

    201

    6

  • © 2012 Landes Bioscience.

    Do not distribute.

    www.landesbioscience.com Cell Cycle 2805

    Mantel-Cox test was used to determine statistical significance.

    Conclusion

    Recent data show that Dicer1 is an essen-tial factor at different stages of normal hematopoiesis. A limitation of the Dicer1-deletion models is the global depletion of miRNAs that presumably results in dis-ruption of many cellular pathways simulta-neously, which hampers the identification of the functions of individual miRNAs. Although some studies show evidence for only a few miRNAs making dominant contributions, such as miR-17~92 in B-cell development, this may be different for other cell types or even be developmental stage-dependent. Overall, Dicer1 mainly controls survival and expansion at the early stages of lymphoid development and controls cellular activities at the terminal maturation stage. The function of Dicer1 is different in myelopoiesis at the earliest developmental stage as Dicer1 is not essen-tial for cell viability but instead controls essential steps in switching from the stem cell stage toward myeloid lineage devel-opment. Although the functions of some miRNAs, such as miR-17/20/93/106 and miR-223, are well-described in immature and mature myeloid cells, respectively,56,63 the miRNA-controlled pathways that are involved at different stages of myelo-poiesis are still largely elusive. Therefore,

    embryonic day (E) 13.5. Genotyping of Dicer1; p53; C/ebpa-Cre;R26-LSL-Eyfp embryos was performed by PCR assays of DNA from tail or foot biopsies. Sequences of primers are available upon request. All primers were obtained from Biolegio BV. For transplantation, 8-week-old recipient mice C57Bl/6, (Jackson Laboratories) were irradiated (8.5 Gy) and tail-vein injected with fetal liver single-cell suspen-sions. Typically, cells from each fetal liver were transplanted into two recipient mice. Tumorigenicity was subsequently moni-tored by daily examination of the trans-planted mice. Mice were euthanized when moribund. All animal experiments were approved by the Animal Welfare/Ethics Committee of the Erasmus Medical Center.

    Antibodies, cell staining, flow cytom-etry and cytospins. Peripheral blood was obtained by heart puncture at the moment of euthanasia. Bone marrow cell suspen-sions were prepared as described previ-ously.34 Tumor samples were prepared as single-cell suspension for cytospins or FACS analysis. For morphological analy-sis of the cells, cytospins were stained with May-Grünwald-Giemsa and examined with a Leica DMLB microscope (100x and 40x objectives) and Leica Application Suite software version 2.7.1 R1.

    Statistics. Kaplan-Meier survival curves were plotted using SPSS software (SPSS, PASW, 17.0.2), and log-rank

    outcome were found in human AML.53 However, there is evidence for regulation of DICER1 expression by miRNAs such as miR-15a and miR-16 in a cohort of del(13q14) in CLL,52 miR-9 in Hodgkin lymphoma,54 miR-125 in human mega-karyoblastic leukemia55 and miR-106a in the undifferentiated primary monocytes.35 Interestingly, miR-9, miR-125 and miR-106a are frequently aberrantly expressed at high levels in human AML44,56 (and review43) and may control DICER1 trans-lation, leaving mRNA levels intact. Thus, aberrant miRNA biogenesis in human AML may occur via direct miRNA-con-trolled feedback mechanisms on trans-lation of DICER1 transcripts, but this hypothesis still needs proper experimental confirmation.

    Reduction of miRNA expression may be controlled by other mechanisms as well. This hypothesis is supported by recently described mutations in the TAR RNA-binding protein 2 (TARBP2), a critical protein for processing miRNAs in sporadic and hereditary carcinomas, and the inactivating mutations in Exportin-5, which results in trap of pre-miRNAs in the nucleus in human cancer cells.57-59 Other possible mechanisms behind aber-rant miRNA expression are single nucleo-tide polymorphisms (SNPs) that influence processing of miRNAs60 or RNA editing of miRNA precursors that blocks cleav-age by DICER1.61,62 Sequencing of factors involved in the biogenesis of miRNAs or a better understanding of miRNA expres-sion regulation by, e.g., transcription fac-tors, epigenetic events or miRNA stability, are needed to unravel the mechanisms behind the reduced miRNA activity in human AML.

    Methods

    Mice and reconstitution experiments. To generate the different mouse lines of interest, we first crossed C/ebpa-Cre;R26-LSL-Eyfp;Dicer1wt/fl mice34 with mice that contain floxed p53 conditional alleles (Jackson Laboratories). Finally, C/ebpa-Cre ;R26-LSL-Eyfp;Dicer1wt / f l /Dicer1fl/ fl;p53 fl/fl mice were obtained from breeding C/ebpa-Cre;Dicer1wt/fl; p53fl/wt mice with R26-LSL-Eyfp;Dicer1fl/fl;p53 fl/fl mice. Fetal livers were obtained on

    Figure 4. Model for the role of Dicer1 in leukemia development. Dicer1 knockout and as a result total loss of mirNA biogenesis, lead to myeloid dysplasia but not leukemia in a p53 knockout background. in contrast, heterozygous loss of Dicer1 conserves the expression of a set of mirNAs needed for normal differentiation. Furthermore, our model suggests that at least some mirNA activity is needed for oncogenic transformation.

    Dow

    nloa

    ded

    by [

    Era

    smus

    Uni

    vers

    ity]

    at 0

    0:53

    17

    June

    201

    6

  • © 2012 Landes Bioscience.

    Do not distribute.

    2806 Cell Cycle volume 11 issue 15

    26. Koralov SB, Muljo SA, Galler GR, Krek A, Chakraborty T, Kanellopoulou C, et al. Dicer abla-tion affects antibody diversity and cell survival in the B lymphocyte lineage. Cell 2008; 132:860-74; PMID:18329371; http://dx.doi.org/10.1016/j.cell.2008.02.020.

    27. Ventura A, Young AG, Winslow MM, Lintault L, Meissner A, Erkeland SJ, et al. Targeted deletion reveals essential and overlapping functions of the miR-17 through 92 family of miRNA clusters. Cell 2008; 132:875-86; PMID:18329372; http://dx.doi.org/10.1016/j.cell.2008.02.019.

    28. Belver L, de Yébenes VG, Ramiro AR. MicroRNAs prevent the generation of autoreactive antibodies. Immunity 2010; 33:713-22; PMID:21093320; http://dx.doi.org/10.1016/j.immuni.2010.11.010.

    29. Xu S, Guo K, Zeng Q, Huo J, Lam KP. The RNase III enzyme Dicer is essential for germinal center B-cell formation. Blood 2012; 119:767-76; PMID:22117047; http://dx.doi.org/10.1182/blood-2011-05-355412.

    30. Bezman NA, Cedars E, Steiner DF, Blelloch R, Hesslein DG, Lanier LL. Distinct requirements of microRNAs in NK cell activation, survival, and function. J Immunol 2010; 185:3835-46; PMID:20805417; http://dx.doi.org/10.4049/jimmu-nol.1000980.

    31. Sullivan RP, Leong JW, Schneider SE, Keppel CR, Germino E, French AR, et al. MicroRNA-deficient NK cells exhibit decreased survival but enhanced function. J Immunol 2012; 188:3019-30; PMID:22379033; http://dx.doi.org/10.4049/jim-munol.1102294.

    32. Guo S, Lu J, Schlanger R, Zhang H, Wang JY, Fox MC, et al. MicroRNA miR-125a controls hema-topoietic stem cell number. Proc Natl Acad Sci USA 2010; 107:14229-34; PMID:20616003; http://dx.doi.org/10.1073/pnas.0913574107.

    33. de Boer J, Williams A, Skavdis G, Harker N, Coles M, Tolaini M, et al. Transgenic mice with hema-topoietic and lymphoid specific expression of Cre. Eur J Immunol 2003; 33:314-25; PMID:12548562; http://dx.doi.org/10.1002/immu.200310005.

    34. Alemdehy MF, van Boxtel NG, de Looper HW, van den Berge IJ, Sanders MA, Cupedo T, et al. Dicer1 deletion in myeloid-committed progenitors causes neutrophil dysplasia and blocks macrophage/dendrit-ic cell development in mice. Blood 2012; 119:4723-30; PMID:22353998; http://dx.doi.org/10.1182/blood-2011-10-386359.

    35. Coley W, Van Duyne R, Carpio L, Guendel I, Kehn-Hall K, Chevalier S, et al. Absence of DICER in monocytes and its regulation by HIV-1. J Biol Chem 2010; 285:31930-43; PMID:20584909; http://dx.doi.org/10.1074/jbc.M110.101709.

    36. Klase Z, Kale P, Winograd R, Gupta MV, Heydarian M, Berro R, et al. HIV-1 TAR element is processed by Dicer to yield a viral micro-RNA involved in chromatin remodeling of the viral LTR. BMC Mol Biol 2007; 8:63; PMID:17663774; http://dx.doi.org/10.1186/1471-2199-8-63.

    37. Lu J, Getz G, Miska EA, Alvarez-Saavedra E, Lamb J, Peck D, et al. MicroRNA expression profiles classify human cancers. Nature 2005; 435:834-8; PMID:15944708; http://dx.doi.org/10.1038/nature03702.

    38. Kumar MS, Lu J, Mercer KL, Golub TR, Jacks T. Impaired microRNA processing enhances cel-lular transformation and tumorigenesis. Nat Genet 2007; 39:673-7; PMID:17401365; http://dx.doi.org/10.1038/ng2003.

    39. Kumar MS, Pester RE, Chen CY, Lane K, Chin C, Lu J, et al. Dicer1 functions as a haploinsuffi-cient tumor suppressor. Genes Dev 2009; 23:2700-4; PMID:19903759; http://dx.doi.org/10.1101/gad.1848209.

    11. Benhamed M, Herbig U, Ye T, Dejean A, Bischof O. Senescence is an endogenous trigger for microRNA-directed transcriptional gene silencing in human cells. Nat Cell Biol 2012; 14:266-75; PMID:22366686; http://dx.doi.org/10.1038/ncb2443.

    12. Reinhart BJ, Weinstein EG, Rhoades MW, Bartel B, Bartel DP. MicroRNAs in plants. Genes Dev 2002; 16:1616-26; PMID:12101121; http://dx.doi.org/10.1101/gad.1004402.

    13. Wienholds E, Koudijs MJ, van Eeden FJ, Cuppen E, Plasterk RH. The microRNA-producing enzyme Dicer1 is essential for zebrafish development. Nat Genet 2003; 35:217-8; PMID:14528306; http://dx.doi.org/10.1038/ng1251.

    14. Bernstein E, Kim SY, Carmell MA, Murchison EP, Alcorn H, Li MZ, et al. Dicer is essential for mouse development. Nat Genet 2003; 35:215-7; PMID:14528307; http://dx.doi.org/10.1038/ng1253.

    15. Kanellopoulou C, Muljo SA, Kung AL, Ganesan S, Drapkin R, Jenuwein T, et al. Dicer-deficient mouse embryonic stem cells are defective in differentiation and centromeric silencing. Genes Dev 2005; 19:489-501; PMID:15713842; http://dx.doi.org/10.1101/gad.1248505.

    16. Murchison EP, Partridge JF, Tam OH, Cheloufi S, Hannon GJ. Characterization of Dicer-deficient murine embryonic stem cells. Proc Natl Acad Sci USA 2005; 102:12135-40; PMID:16099834; http://dx.doi.org/10.1073/pnas.0505479102.

    17. Harfe BD, McManus MT, Mansfield JH, Hornstein E, Tabin CJ. The RNaseIII enzyme Dicer is required for morphogenesis but not patterning of the vertebrate limb. Proc Natl Acad Sci USA 2005; 102:10898-903; PMID:16040801; http://dx.doi.org/10.1073/pnas.0504834102.

    18. Cobb BS, Nesterova TB, Thompson E, Hertweck A, O’Connor E, Godwin J, et al. T cell lineage choice and differentiation in the absence of the RNase III enzyme Dicer. J Exp Med 2005; 201:1367-73; PMID:15867090; http://dx.doi.org/10.1084/jem.20050572.

    19. Muljo SA, Ansel KM, Kanellopoulou C, Livingston DM, Rao A, Rajewsky K. Aberrant T cell dif-ferentiation in the absence of Dicer. J Exp Med 2005; 202:261-9; PMID:16009718; http://dx.doi.org/10.1084/jem.20050678.

    20. Cobb BS, Hertweck A, Smith J, O’Connor E, Graf D, Cook T, et al. A role for Dicer in immune regulation. J Exp Med 2006; 203:2519-27; PMID:17060477; http://dx.doi.org/10.1084/jem.20061692.

    21. Liston A, Lu LF, O’Carroll D, Tarakhovsky A, Rudensky AY. Dicer-dependent microRNA pathway safeguards regulatory T cell function. J Exp Med 2008; 205:1993-2004; PMID:18725526; http://dx.doi.org/10.1084/jem.20081062.

    22. Zhou X, Jeker LT, Fife BT, Zhu S, Anderson MS, McManus MT, et al. Selective miRNA disruption in T reg cells leads to uncontrolled autoimmunity. J Exp Med 2008; 205:1983-91; PMID:18725525; http://dx.doi.org/10.1084/jem.20080707.

    23. Zhou L, Seo KH, He HZ, Pacholczyk R, Meng DM, Li CG, et al. Tie2cre-induced inactivation of the miRNA-processing enzyme Dicer disrupts invariant NKT cell development. Proc Natl Acad Sci USA 2009; 106:10266-71; PMID:19509335; http://dx.doi.org/10.1073/pnas.0811119106.

    24. Batard P, Sansilvestri P, Scheinecker C, Knapp W, Debili N, Vainchenker W, et al. The Tie receptor tyrosine kinase is expressed by human hematopoietic progenitor cells and by a subset of megakaryocytic cells. Blood 1996; 87:2212-20; PMID:8630381.

    25. Fedeli M, Napolitano A, Wong MP, Marcais A, de Lalla C, Colucci F, et al. Dicer-dependent microRNA pathway controls invariant NKT cell development. J Immunol 2009; 183:2506-12; PMID:19625646; http://dx.doi.org/10.4049/jimmunol.0901361.

    tissue and developmental stage-specific miRNA-add-back in the Dicer1-deficient models and experimental target identifi-cation approaches may be of help for the understanding of the miRNA activities in hematopoiesis.

    Acknowledgments

    We thank Dr. T Cupedo and Dr. M. Buitenhuis for critical reading of the man-uscript and E. Simons for assistance with the preparation of the figures. We also thank Dr. K. van Lom for the microscopic analysis of leukemias. Our work was sup-ported by grants from the Netherlands Organisation for Scientific Research (NWO-VENI) and the Dutch Cancer Society (KWF). There are no conflicts of interests.

    References1. Dlakic M. DUF283 domain of Dicer proteins has a

    double-stranded RNA-binding fold. Bioinformatics 2006; 22:2711-4; PMID:16954143; http://dx.doi.org/10.1093/bioinformatics/btl468.

    2. Qin H, Chen F, Huan X, Machida S, Song J, Yuan YA. Structure of the Arabidopsis thaliana DCL4 DUF283 domain reveals a noncanonical double-stranded RNA-binding fold for protein-protein inter-action. RNA 2010; 16:474-81; PMID:20106953; http://dx.doi.org/10.1261/rna.1965310.

    3. Lau PW, Guiley KZ, De N, Potter CS, Carragher B, MacRae IJ. The molecular architecture of human Dicer. Nat Struct Mol Biol 2012; 19:436-40; PMID:22426548; http://dx.doi.org/10.1038/nsmb.2268.

    4. Bernstein E, Caudy AA, Hammond SM, Hannon GJ. Role for a bidentate ribonuclease in the ini-tiation step of RNA interference. Nature 2001; 409:363-6; PMID:11201747; http://dx.doi.org/10.1038/35053110.

    5. Calabrese JM, Seila AC, Yeo GW, Sharp PA. RNA sequence analysis defines Dicer’s role in mouse embryonic stem cells. Proc Natl Acad Sci USA 2007; 104:18097-102; PMID:17989215; http://dx.doi.org/10.1073/pnas.0709193104.

    6. Cheloufi S, Dos Santos CO, Chong MM, Hannon GJ. A dicer-independent miRNA biogenesis pathway that requires Ago catalysis. Nature 2010; 465:584-9; PMID:20424607; http://dx.doi.org/10.1038/nature09092.

    7. Cifuentes D, Xue H, Taylor DW, Patnode H, Mishima Y, Cheloufi S, et al. A novel miRNA processing pathway independent of Dicer requires Argonaute2 catalytic activity. Science 2010; 328:1694-8; PMID:20448148; http://dx.doi.org/10.1126/science.1190809.

    8. Yang JS, Maurin T, Robine N, Rasmussen KD, Jeffrey KL, Chandwani R, et al. Conserved vertebrate mir-451 provides a platform for Dicer-independent, Ago2-mediated microRNA biogenesis. Proc Natl Acad Sci USA 2010; 107:15163-8; PMID:20699384; http://dx.doi.org/10.1073/pnas.1006432107.

    9. Yang JS, Lai EC. Dicer-independent, Ago2-mediated microRNA biogenesis in vertebrates. Cell Cycle 2010; 9:4455-60; PMID:21088485; http://dx.doi.org/10.4161/cc.9.22.13958.

    10. Ketting RF. The many faces of RNAi. Dev Cell 2011; 20:148-61; PMID:21316584; http://dx.doi.org/10.1016/j.devcel.2011.01.012.

    Dow

    nloa

    ded

    by [

    Era

    smus

    Uni

    vers

    ity]

    at 0

    0:53

    17

    June

    201

    6

  • © 2012 Landes Bioscience.

    Do not distribute.

    www.landesbioscience.com Cell Cycle 2807

    56. Meenhuis A, van Veelen PA, de Looper H, van Boxtel N, van den Berge IJ, Sun SM, et al. MiR-17/20/93/106 promote hematopoietic cell expansion by targeting sequestosome 1-regulated pathways in mice. Blood 2011; 118:916-25; PMID:21628417; http://dx.doi.org/10.1182/blood-2011-02-336487.

    57. Melo SA, Ropero S, Moutinho C, Aaltonen LA, Yamamoto H, Calin GA, et al. A TARBP2 mutation in human cancer impairs microRNA processing and DICER1 function. Nat Genet 2009; 41:365-70; PMID:19219043; http://dx.doi.org/10.1038/ng.317.

    58. Melo SA, Moutinho C, Ropero S, Calin GA, Rossi S, Spizzo R, et al. A genetic defect in exportin-5 traps precursor microRNAs in the nucleus of cancer cells. Cancer Cell 2010; 18:303-15; PMID:20951941; http://dx.doi.org/10.1016/j.ccr.2010.09.007.

    59. Melo SA, Esteller M. A precursor microRNA in a cancer cell nucleus: get me out of here! Cell Cycle 2011; 10:922-5; PMID:21346411; http://dx.doi.org/10.4161/cc.10.6.15119.

    60. Sun G, Yan J, Noltner K, Feng J, Li H, Sarkis DA, et al. SNPs in human miRNA genes affect biogenesis and function. RNA 2009; 15:1640-51; PMID:19617315; http://dx.doi.org/10.1261/rna.1560209.

    61. Kawahara Y, Zinshteyn B, Chendrimada TP, Shiekhattar R, Nishikura K. RNA editing of the microRNA-151 precursor blocks cleavage by the Dicer-TRBP complex. EMBO Rep 2007; 8:763-9; PMID:17599088; http://dx.doi.org/10.1038/sj.embor.7401011.

    62. Heale BS, Keegan LP, O’Connell MA. ADARs have effects beyond RNA editing. Cell Cycle 2009; 8:4011-2; PMID:19949296; http://dx.doi.org/10.4161/cc.8.24.10214.

    63. Johnnidis JB, Harris MH, Wheeler RT, Stehling-Sun S, Lam MH, Kirak O, et al. Regulation of progenitor cell proliferation and granulocyte func-tion by microRNA-223. Nature 2008; 451:1125-9; PMID:18278031; http://dx.doi.org/10.1038/nature06607.

    48. Zhang H, Kolb FA, Jaskiewicz L, Westhof E, Filipowicz W. Single processing center models for human Dicer and bacterial RNase III. Cell 2004; 118:57-68; PMID:15242644; http://dx.doi.org/10.1016/j.cell.2004.06.017.

    49. Gurtan AM, Lu V, Bhutkar A, Sharp PA. In vivo structure-function analysis of human Dicer reveals directional processing of precursor miRNAs. RNA 2012; 18:1116-22; PMID:22546613; http://dx.doi.org/10.1261/rna.032680.112.

    50. Deng S, Calin GA, Croce CM, Coukos G, Zhang L. Mechanisms of microRNA deregulation in human cancer. Cell Cycle 2008; 7:2643-6; PMID:18719391; http://dx.doi.org/10.4161/cc.7.17.6597.

    51. Merritt WM, Lin YG, Han LY, Kamat AA, Spannuth WA, Schmandt R, et al. Dicer, Drosha, and out-comes in patients with ovarian cancer. N Engl J Med 2008; 359:2641-50; PMID:19092150; http://dx.doi.org/10.1056/NEJMoa0803785.

    52. Zhu DX, Fan L, Lu RN, Fang C, Shen WY, Zou ZJ, et al. Downregulated Dicer expression predicts poor prognosis in chronic lymphocytic leukemia. Cancer Sci 2012; 103:875-81; PMID:22320315; http://dx.doi.org/10.1111/j.1349-7006.2012.02234.x.

    53. Martin MG, Payton JE, Link DC. Dicer and out-comes in patients with acute myeloid leukemia (AML). Leuk Res 2009; 33:e127; PMID:19278725; http://dx.doi.org/10.1016/j.leukres.2009.02.003.

    54. Leucci E, Zriwil A, Gregersen LH, Jensen KT, Obad S, Bellan C, et al. Inhibition of miR-9 de-represses HuR and DICER1 and impairs Hodgkin lymphoma tumour outgrowth in vivo. Oncogene 2012; PMID:22310293; http://dx.doi.org/10.1038/onc.2012.15.

    55. Klusmann JH, Li Z, Böhmer K, Maroz A, Koch ML, Emmrich S, et al. miR-125b-2 is a poten-tial oncomiR on human chromosome 21 in mega-karyoblastic leukemia. Genes Dev 2010; 24:478-90; PMID:20194440; http://dx.doi.org/10.1101/gad.1856210.

    40. Lambertz I, Nittner D, Mestdagh P, Denecker G, Vandesompele J, Dyer MA, et al. Monoallelic but not biallelic loss of Dicer1 promotes tumorigen-esis in vivo. Cell Death Differ 2010; 17:633-41; PMID:20019750; http://dx.doi.org/10.1038/cdd.2009.202.

    41. Arrate MP, Vincent T, Odvody J, Kar R, Jones SN, Eischen CM. MicroRNA biogenesis is required for Myc-induced B-cell lymphoma development and survival. Cancer Res 2010; 70:6083-92; PMID:20587524; http://dx.doi.org/10.1158/0008-5472.CAN-09-4736.

    42. Wölfler A, Danen-van Oorschot AA, Haanstra JR, Valkhof M, Bodner C, Vroegindeweij E, et al. Lineage-instructive function of C/EBPα in multipo-tent hematopoietic cells and early thymic progenitors. Blood 2010; 116:4116-25; PMID:20807890; http://dx.doi.org/10.1182/blood-2010-03-275404.

    43. Alemdehy MF, Erkeland SJ. MicroRNAs: key players of normal and malignant myelopoiesis. Curr Opin Hematol 2012; 19:261-7; PMID:22504525; http://dx.doi.org/10.1097/MOH.0b013e328353d4e9.

    44. Jongen-Lavrencic M, Sun SM, Dijkstra MK, Valk PJ, Löwenberg B. MicroRNA expression profiling in relation to the genetic heterogeneity of acute myeloid leukemia. Blood 2008; 111:5078-85; PMID:18337557; http://dx.doi.org/10.1182/blood-2008-01-133355.

    45. Forbes SA, Bhamra G, Bamford S, Dawson E, Kok C, Clements J, et al. The Catalogue of Somatic Mutations in Cancer (COSMIC). Current protocols in human genetics / editorial board, Jonathan L Haines [et al 2008; Chapter 10:Unit 10 1.

    46. Hill DA, Ivanovich J, Priest JR, Gurnett CA, Dehner LP, Desruisseau D, et al. DICER1 muta-tions in familial pleuropulmonary blastoma. Science 2009; 325:965; PMID:19556464; http://dx.doi.org/10.1126/science.1174334.

    47. Heravi-Moussavi A, Anglesio MS, Cheng SW, Senz J, Yang W, Prentice L, et al. Recurrent somatic DICER1 mutations in nonepithelial ovarian cancers. N Engl J Med 2012; 366:234-42; PMID:22187960; http://dx.doi.org/10.1056/NEJMoa1102903.

    Dow

    nloa

    ded

    by [

    Era

    smus

    Uni

    vers

    ity]

    at 0

    0:53

    17

    June

    201

    6


Recommended